EP3978031A1 - Thérapies à base de transcriptase inverses de la télomérase - Google Patents

Thérapies à base de transcriptase inverses de la télomérase Download PDF

Info

Publication number
EP3978031A1
EP3978031A1 EP21198809.2A EP21198809A EP3978031A1 EP 3978031 A1 EP3978031 A1 EP 3978031A1 EP 21198809 A EP21198809 A EP 21198809A EP 3978031 A1 EP3978031 A1 EP 3978031A1
Authority
EP
European Patent Office
Prior art keywords
vector
tert
seq
viral vector
use according
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21198809.2A
Other languages
German (de)
English (en)
Inventor
Maria Bobadilla
Ivan FORMENTINI
Maria Antonia Blasco Marhuenda
Christian Baer
Fatima Bosch Tubert
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universitat Autonoma de Barcelona UAB
Fundacion del Sector Publico Estatal Centro Nacional de Investigaciones Oncologicas Carlos III FSP CNIO
Original Assignee
Universitat Autonoma de Barcelona UAB
Fundacion del Sector Publico Estatal Centro Nacional de Investigaciones Oncologicas Carlos III FSP CNIO
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universitat Autonoma de Barcelona UAB, Fundacion del Sector Publico Estatal Centro Nacional de Investigaciones Oncologicas Carlos III FSP CNIO filed Critical Universitat Autonoma de Barcelona UAB
Publication of EP3978031A1 publication Critical patent/EP3978031A1/fr
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0271Chimeric vertebrates, e.g. comprising exogenous cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0058Nucleic acids adapted for tissue specific expression, e.g. having tissue specific promoters as part of a contruct
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • A61K48/005Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy characterised by an aspect of the 'active' part of the composition delivered, i.e. the nucleic acid delivered
    • A61K48/0066Manipulation of the nucleic acid to modify its expression pattern, e.g. enhance its duration of expression, achieved by the presence of particular introns in the delivered nucleic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/12Transferases (2.) transferring phosphorus containing groups, e.g. kinases (2.7)
    • C12N9/1241Nucleotidyltransferases (2.7.7)
    • C12N9/1276RNA-directed DNA polymerase (2.7.7.49), i.e. reverse transcriptase or telomerase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y207/00Transferases transferring phosphorus-containing groups (2.7)
    • C12Y207/07Nucleotidyltransferases (2.7.7)
    • C12Y207/07049RNA-directed DNA polymerase (2.7.7.49), i.e. telomerase or reverse-transcriptase
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/12Animals modified by administration of exogenous cells
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/20Animal model comprising regulated expression system
    • A01K2217/206Animal model comprising tissue-specific expression system, e.g. tissue specific expression of transgene, of Cre recombinase
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0381Animal model for diseases of the hematopoietic system
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14145Special targeting system for viral vectors

Definitions

  • This invention falls within the field of molecular biology, biotechnology and medicine. More particularly, it relates to compositions and methods useful for the treatment of conditions associated with short telomere length. More particularly, it relates to compositions and methods useful for the treatment of conditions associated with aplastic anemia.
  • telomeres are specialized structures at the ends of chromosomes, which have a role in protecting the chromosome ends from DNA repair and degrading activities ( Blackburn, 2001. Cell 106, 661-673 ; de Lange, 2005. Genes Dev. 19, 2100- 2110 ).
  • Mammalian telomeres consist of TTAGGG repeats bound by a multi-protein complex known as shelterin ( de Lange, 2005. Genes Dev. 19, 2100-2110 ). A minimum length of TTAGGG repeats and the integrity of the shelterin complex are necessary for telomere protection ( Blackburn, 2001. Cell 106, 661-673 ; de Lange, 2005. Genes Dev. 19, 2100-2110 ).
  • Telomerase is a cellular reverse transcriptase (TERT, telomerase reverse transcriptase; also known as TP2; TRT; EST2; TCS1; hEST2) capable of compensating telomere attrition through de novo addition of TTAGGG repeats onto the chromosome ends by using an associated R A component as template (Terc, telomerase RNA component) ( Greider and Blackburn, 1985. Cell 43, 405-413 ). Telomerase is expressed in most adult stem cell compartments, however, this is not sufficient to maintain telomere length as evidenced by the fact that telomere shortening occurs with age in most human and mouse tissues ( Harley et al., 1990. Nature 345, 458- 460 ; Blasco, 2007. Nat Chem Biol. 3, 640-649 ; Flores et al, 2008. Genes and Dev 22, 654-667 ).
  • TERT telomerase reverse transcriptase
  • mice carrying homozygous deletion for the TERC gene lack any detectable telomerase activity and showed progressive telomere shortening from one generation to the other at a rate comparable to the rate reported in human cells (Blasco et al., 1997). Severe phenotypes typical of late generation TERC-/- mice (e.g. bone marrow aplasia and signs of premature aging) could be rescued by re-introducing a copy of the TERC gene (Samper et al., 2001).
  • a virus (AAV) based telomerase gene therapy was found to be beneficial to extend health span, in the context of normal physiological aging in wild-type mice.
  • AAV9-mTERT gene therapy was significantly increased, and aging was decelerated, as indicated by a number of physiological parameters (glucose and insulin tolerance, osteoporosis, neuromuscular coordination, rota-rod, etc).
  • their mean lifespan, compared to control groups was increased by 24% and 13% in adult an old mice, respectively.
  • a single intravenous administration of AAV9-TERT in adult mice resulted in an increase in telomere length in peripheral blood cells (Bernardes de Jesus et al., 2012).
  • Shortened telomeres have been associated with numerous diseases, such as Dyskeratosis congenita, Aplastic anaemia, Myelodysplastic Syndrome, and Fanconi anaemia. Given the severity of these diseases and the poor prognosis of the patients suffering from them, there is a need for novel therapies to treat diseases associated with short telomere length.
  • Aplastic anemia is a potentially life-threatening, rare and heterogeneous disorder of the blood in which the bone marrow cannot produce sufficiently enough new blood cells due to a marked reduction of immature hematopoietic stem (HSC) and progenitor cells (Scopes et al., 1994, Maciejewski et al., 1994). Accordingly, the main disease manifestations are pancytopenia and marrow hypoplasia which can emerge at any stage of life but are more frequent in young people (age 10-25years) and the elderly (>60years) (Marsh et al., 2009). Aplastic anemia can be acquired or inherited.
  • the acquired type is mainly autoimmune-mediated but can also be triggered by environmental factors such as radiation, toxin and virus exposure (Nakao, 1997).
  • the congenital form is rarer, however, mutations in more than 30 genes with functions in DNA repair, ribosome biogenesis and telomere maintenance pathways have been identified to date (Dokal & Vulliamy, 2010).
  • a frequently observed clinical feature of aplastic anemia is short telomere length in peripheral blood leukocytes even in the absence of mutations in the telomere maintenance machinery.
  • Telomeres the termini of vertebrate chromosomes are highly specialised nucleoprotein structures composed of hexanucleotide (TTAGGG) tandem repeat sequences which are bound by a six protein complex ( TRF1, TRF2, TIN2, RAP1, TPP1 and POT1 ) termed shelterin (Blackburn, 2001, de Lange, 2005). These structures are essential for chromosome integrity by preventing telomere fusions and telomere fragility. Telomere length is controlled by the ribonucleoprotein enzyme telomerase which can de novo add telomeric sequences onto telomeres.
  • telomeres Because telomeric sequence is naturally lost upon every cell division (known as the end replication problem) and somatic cells express telomerase at very low levels or not at all telomeres shorten throughout life. When telomeres become critically short they lose their protective function and a persistent DNA damage response at the telomeres is triggered which subsequently leads to a cellular senescence response (Harley et al., 1990, Flores et al., 2008). HSCs, in contrast to most somatic cells, show low level of telomerase activity. However, this activity is insufficient to stop telomere attrition and consequently the regeneration potential of HSCs cells may become limited during the aging process (Hiyama & Hiyama, 2007).
  • telomere lengths In line with this, recipients of bone marrow transplants have shorter telomere lengths than their donors suggesting that telomerase cannot cope with increased replicative proliferation demand during the engraftment phase (Wynn et al., 1998). Moreover, telomeres have been shown to shorten much faster in patients with aplastic anemia compared to the normal aging-related attrition found in healthy individuals potentially owed to a higher than normal number of cell divisions (Ball et al., 1998).
  • tissues with a high proliferative index such as the hematopoietic system are particularly affected by lower than normal telomerase levels which can ultimately lead to severe disorders such as aplastic anemia (Vulliamy et al., 2002).
  • telomeropathy dyskeratosis congenita has been linked to mutations in 7 genes with important functions in telomere maintenance ( TERT, TERC, DKC1, TIN2, NOP10, NHP2 and TCAB1 ) and is characterized by very short telomeres.
  • Dyskeratosis congenita is a multisystem syndrome comprising diverse clinical features such as nail dystrophy, oral leucoplakia, abnormal skin pigmentation and cerebellar hypoplasia (Dokal, 2011).
  • telomere length The causality between proliferation potential and telomere length suggests that a therapeutic intervention with telomerase, aimed at preventing telomere loss beyond a critically short length, may be a feasible strategy to treat those forms of aplastic anemia associated with limited blood forming capacity due to the presence of short telomeres.
  • telomerase aimed at preventing telomere loss beyond a critically short length
  • AAV9 adeno-associated virus
  • telomerase gene therapy using AAV9 Tert in adult wilt-type mice attenuated or reverted the aging-associated telomere erosion in peripheral blood monocytes (Bernardes de Jesus et al., 2012), suggesting that this gene therapy may be effective in the treatment of hematological disorders related to short telomeres.
  • Trf1 the shelterin gene
  • Trf1 cause severe telomere uncapping and provokes a DNA damage response which in turn leads to a fast clearance of those HSCs and progenitor cells deficient for Trf1.
  • Trf1 deletion at a frequency that does not target 100% of the HSCs and progenitor cells. Therefore, cells that retain intact Trf1 undergo additional rounds of compensatory proliferation leading to fast telomere attrition.
  • Trf1 deletion recapitulates the compensatory hyperproliferation observed after bone marrow transplantation or in autoimmune-mediated aplastic anemia, as well as presence of very short telomeres in patients owing to mutations in telomere maintenance genes.
  • Trf1 deletion-mediated HSC depletion allows to control the onset of bone marrow aplasia and pancytopenia (Beier et al., 2012).
  • telomere activation using state of the art gene therapy vectors can be an effective treatment to attenuate telomere attrition and HSC depletion, and thus prevent bone marrow failure.
  • the invention provides compositions and methods useful for the treatment and prevention of conditions associated with short telomere length.
  • One aspect of the invention provides a method of treating a patient with a condition associated with short telomere length comprising administering to the patient a nucleic acid vector comprising a coding sequence for telomerase reverse transcriptase (TERT).
  • TERT telomerase reverse transcriptase
  • the TERT is encoded by a nucleic acid sequence comprising a sequence that is at least 90% identical to the sequence of SEQ ID NO: 1 or SEQ ID NO: 3.
  • the TERT is encoded by a nucleic acid sequence comprising the sequence of SEQ ID NO: 1 or SEQ ID NO: 3.
  • the TERT is encoded by a nucleic acid sequence consisting of the sequence of SEQ ID NO: 1 or SEQ ID NO: 3
  • the TERT comprises an amino acid sequence that is at least 90% identical to the amino acid sequence of SEQ ID NO:2 or SEQ ID NO: 4.
  • the TERT comprises an amino acid sequence of SEQ ID NO:2 or SEQ ID NO: 4.
  • the TERT consists of the amino acid sequence of SEQ ID NO:2 or SEQ ID NO: 4.
  • the nucleic acid sequence encoding TERT is operably linked to a regulatory sequence that drives the expression of the coding sequence.
  • the vector is a non-integrative vector, such as an adeno-associated virus-based non-integrative vector.
  • the vector is an adeno-associated virus-based vector derived from a serotype 9 adeno-associated virus (AAV9).
  • the capsid of the adeno-associated virus-based vector is made of capsid proteins of the serotype 9 adeno-associated virus (AAV9), and the nucleic acid sequence contained in the capsid is flanked at both ends by internal terminal repeats corresponding to serotype 2 adenoassociated viruses.
  • the nucleic acid contained in the capsid comprises a fragment which encodes the amino acid sequence coding for TERT.
  • the vector comprises a regulatory sequence which is a constitutive promoter.
  • the regulatory sequence is the cytomegalovirus (CMV) promoter.
  • CMV cytomegalovirus
  • the condition associated with short telomere length is characterized by mutations in a gene or genes involved in telomere maintenance.
  • the condition associated with short telomere length is selected from the group consisting of Dyskeratosis congenita, Aplastic anaemia, Myelodysplastic Syndrome, Fanconi anaemia.
  • the invention is directed to the following set of subject matters:
  • the invention provides compositions and methods useful for the treatment and prevention of conditions associated with short telomere length.
  • a "condition associated with short telomere length” is one which is characterized by an accumulation of critically short telomeres.
  • subjects suffering from such a condition exhibit premature onset of pathologies resulting from a defective regenerative capacity of tissues.
  • the condition associated with short telomere length is characterized by mutations in a gene or genes involved in telomere maintenance.
  • specific examples of such genetically based conditions include, but are not limited to Dyskeratosis congenita, Aplastic anaemia, Myelodysplastic Syndrome, Fanconi anaemia, and pulmonary fibrosis.
  • Dyskeratosis congenita is a genetically heterogeneous human disease, which is paradigmatic of premature ageing syndromes (Dokal, 2011). DKC is characterised by the presence of short/dysfunctional telomeres owing to mutations in genes related to telomere maintenance, being the most frequently mutated those encoding proteins of the telomerase complex (i.e. TERT, TERC, NOP10, DKC1, NHP2) (Dokal, 2011; Dokal and Vulliamy, 2010; Mason and Bessler, 2011; Savage and Alter, 2008).
  • TIN2 TRF1-interacting protein
  • a component of the shelterin complex which binds and protects mammalian telomeres (Dokal, 2011; Martinez and Blasco, 2011; Walne et al., 2008).
  • a functional telomerase complex and a proper telomere capping structure by the shelterin proteins are required for maintenance and capping of chromosome ends, respectively.
  • Clinical features of patients suffering from DKC include skin abnormalities (i.e. skin hyperpigmentation), signs of premature aging (i.e. hair greying, nail dystrophy, oral leucoplakia, etc), predisposition to cancer, and several other life-threatening conditions, including aplastic anemia and pulmonary fibrosis (Armanios and Blackburn, 2012).
  • tissues with a high proliferative index are most affected due to the loss of telomeric DNA that occurs upon each cell division. This explains why DKC patients are particularly vulnerable to impaired bone marrow function leading to pancytopenia and eventually bone marrow failure (BMF) (Armanios and Blackburn, 2012; Blasco, 2007)
  • Aplastic anaemia is a life threatening bone marrow disorder characterised by hypocellular bone marrow and low blood cell counts. Patients with acquired aplastic anaemia present with leukocytes which have considerably shorter telomeres than age-matched healthy individuals (Carroll and Ly, 2009). Aplastic anaemia is frequently caused by an autoimmune mediated attack against hematopoietic stem cells.
  • mutations in the core telomerase components TERT and TERC are the underlying cause in a clinically relevant subpopulation (Yamaguchi et al., 2003; Yamaguchi et al., 2005). Mutations in the core telomerase components TERT and TERC, as well as in the shelterin component TIN2 have been linked to this disease (Savage et al., 2006).
  • Myelodysplastic Syndrome encompasses several bone marrow diseases characterised by ineffective production of the myeloid class of blood cells.
  • MDS patients often report with severe anaemia and cytopenias.
  • AML acute myelogenous leukemia
  • shortened telomeres in patients with MDS suggest that insufficient or hampered telomeric maintenance is causative for the syndrome, only 3 out of 210 cases showed heterozygous TERC mutations in a previous study (Yamaguchi et al., 2003).
  • Fanconi anaemia is a heterogeneous genetic disease caused by mutations in genes involved in DNA repair. Affected individuals display multiple congenital defects and haematological deficiencies at a young age of onset (Kee and D'Andrea, 2012). Manifestations related to the latter however are the predominant symptoms of this syndrome and as the disease progresses can develop into the aforementioned syndromes including aplastic anaemia, MDS and AML. Importantly, patients suffering from FA have been also shown to present shorter telomeres than normal (Gadalla et al., 2010). The facts that mutations causing FA show impaired DNA damage response (DDR) and telomeres are particularly vulnerable to replicative stress may provide an explanation for the observed telomere erosion. In support of this Callen et al. (2002) suggested that in FA patients increased telomere breakage in concert with replicative shortening account for the observed telomere shortening.
  • DDR DNA damage response
  • Pulmonary fibrosis refers to a condition characterised by scarring of the lung tissue. Pulmonary fibrosis can be caused by many factors, including chronic inflammatory processes, infections, environmental compounds, ionizing radiation (for example radiation therapy to treat tumors of the chest), chronic medical conditions (lupus, rheumatoid arthritis). Idiopathic pulmonary fibrosis (IPF) refers to pulmonary fibrosis without an identifiable cause.
  • IPF Idiopathic pulmonary fibrosis
  • the invention provides methods of treating a patient suffering from a condition associated with short telomere length comprising administering to the patient an agent which increases the telomere length of the patient.
  • the agent prevents degradation of the chromosomal ends.
  • the agent increases the activity of telomerase reverse transcriptase (TERT).
  • the method of treatment is a gene therapy method comprises administering to the patient a nucleic acid vector comprising a coding sequence for telomerase reverse transcriptase (TERT).
  • the TERT sequence used in the gene therapy vector is derived from the same species as the subject.
  • gene therapy in humans would be carried out using the human TERT sequence.
  • Gene therapy in mice would be carried out using the mouse TERT sequence, as described in the examples.
  • the TERT is encoded by the nucleic acid sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 3 (human TERT variants 1 and 2), or is an active fragment or functional equivalent of SEQ ID NO: 1 or SEQ ID NO: 3.
  • the polypeptide sequence encoded by SEQ ID NO: 1 is set forth in SEQ ID NO: 2.
  • the polypeptide encoded by SEQ ID NO: 3 is set forth in SEQ ID NO: 4.
  • “functional equivalent” refers to a nucleic acid molecule that encodes a polypeptide that has TERT activity or a polypeptide that has TERT activity.
  • the functional equivalent may displays 50%, 60%, 70%, 80%, 90%, 95%, 98%, 99%, 100% or more activity compared to TERT encoded by SEQ ID NO: 1 or SEQ ID NO: 3.
  • Functional equivalents may be artificial or naturally-occurring. For example, naturally- occurring variants of the TERT sequence in a population fall within the scope of functional equivalent. TERT sequences derived from other species also fall within the scope of the term "functional equivalent", in particular the murine TERT sequence given in SEQ ID NO: 5.
  • the functional equivalent is a nucleic acid with a nucleotide sequence having at least 75%, 80%>, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9% identity to SEQ ID NO: 1 or SEQ ID NO: 3.
  • the functional equivalent is a polypeptide with an amino acid sequence having at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9% identity to SEQ ID NO: 2 or SEQ ID NO: 4.
  • sequence identity should be calculated along the entire length of the nucleic acid.
  • Functional equivalents may contain one or more, e.g.
  • nucleotide insertions, deletions and/or substitutions when compared to SEQ ID NO: 1 or SEQ ID NO: 3.
  • the term "functional equivalent” also encompasses nucleic acid sequences that encode a TERT polypeptide with at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9% sequence identity to the sequence as set forth in SEQ ID NO:2 or SEQ ID NO: 4, but that show little homology to the nucleic acid sequence given in SEQ ID NO: 1 or SEQ ID NO: 3 because of the degeneracy of the genetic code.
  • active fragment refers to a nucleic acid molecule that encodes a polypeptide that has TERT activity or polypeptide that has TERT activity, but which is a fragment of the nucleic acid as set forth in SEQ ID NO: 1 or SEQ ID NO: 3 or the amino acid sequence as set forth in SEQ ID NO: 2 or SEQ ID NO: 4.
  • An active fragment may be of any size provided that TERT activity is retained.
  • a fragment will have at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 100% identity to SEQ ID NO: 1-4 along the length of the alignment between the shorter fragment and SEQ ID NO: 1-4.
  • Fusion proteins including these fragments can be comprised in the nucleic acid vectors needed to carry out the invention.
  • an additional 5, 10, 20, 30, 40, 50 or even 100 amino acid residues from the polypeptide sequence, or from a homologous sequence may be included at either or both the C terminal and/or N terminus without prejudicing the ability of the polypeptide fragment to fold correctly and exhibit biological activity.
  • Sequence identity may be calculated by any one of the various methods in the art, including for example BLAST ( Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ (1990). "Basic local alignment search tool”. J Mol Biol 215 (3): 403-410 ) and FASTA ( Lipman, DJ; Pearson, WR (1985). "Rapid and sensitive protein similarity searches”. Science 227 (4693): 1435-41; http ://fasta.bioch. Virginia, edu/ fasta_www2/ fasta_list2. shtml ) and variations on these alignment programs.
  • the method of treatment is a gene therapy method and/or the nucleic acid vector used is a gene therapy vector.
  • Gene therapy methods and vectors are well known in the art and generally comprise delivering a nucleic acid encoding a therapeutically active protein to a subject.
  • the nucleic acid may be delivered in a number of ways including delivering naked DNA such as plasmid or mini-circles, the use of liposomes or cationic polymers or other engineered nano-particles containing the nucleic acid, or viral vectors that encapsidate the nucleic acid.
  • the gene therapy is achieved using stable transformation of organisms with an inducible expression system.
  • Suitable inducible expression systems include the CRE-LOX recombinase based system which is suitable for use in mice and tetracycline-regulated which can be used in the treatment of human subjects.
  • the gene therapy vector is a viral vector.
  • Viral gene therapy vectors are well known in the art. Vectors include integrative and non-integrative vectors such as those based on retroviruses, adenoviruses (AdV), adeno- associated viruses (AAV), lentiviruses, pox viruses, alphaviruses, and herpes viruses.
  • non-integrative viral vectors such as AAV
  • AAV a non-integrative viral vectors
  • the vectors target to adult tissues, avoiding having the subjects under the effect of constitutive telomerase expression from early stages of development.
  • non-integrative vectors effectively incorporate a safety mechanism to avoid over-proliferation of TERT expressing cells. Cells will lose the vector (and, as a consequence, the telomerase expression) if they start proliferating quickly.
  • non-integrative vectors include those based on adenoviruses (AdV) in particular gutless adenoviruses, adeno-associated viruses (AAV), integrase deficient lentiviruses , pox viruses, alphaviruses, and herpes viruses.
  • AdV adenoviruses
  • AAV adeno-associated viruses
  • the non-integrative vector used in the invention is an adeno-associated virus- based non-integrative vector, similar to natural adeno-associated virus particles.
  • AAV preferentially targets post-mitotic tissues, which are considered more resistant to cancer than the highly proliferative ones.
  • Examples of adeno-associated virus-based non integrative vectors include vectors based on any AAV serotype, i.e.
  • Tissue specificity is determined by the capsid serotype. Pseudotyping of AAV vectors and capsid engineering to alter their tropism range will likely be important to their use in therapy.
  • AAVs adeno-associated viruses
  • AAV vectors transduce post-mitotic cells and can sustain long-term gene expression (up to several years) both in small and large animal models of disease ( Niemeyer, Herzog et al Blood 2009 ; Mas, Montane et al Diabetes 2006 ; Jiang, Lillicrap et al blood 2006 ; Ghosh, Yue et al Molecular therapy 2007 ; Tafuro, Ayuso et al cardiovascular research 2009 ).
  • AAV2 is the best characterized serotype for gene transfer studies both in humans and experimental models.
  • AAV2 presents natural tropism towards skeletal muscles, neurons, vascular smooth muscle cells and hepatocytes.
  • AAV2 is therefore a good choice of vector to target these tissues, in particular when using the methods or vectors of the invention to treat a condition associated with one of these tissues. For example, treatment of neuromuscular degeneration may be targeted to skeletal muscle and/or neurons in this way.
  • Newly isolated serotypes such as AAV7, AAV8, and AAV9 have been successfully adopted in preclinical studies ( Gao, Alvira et al PNAS 2002 ). Although limited immunologic responses have been detected in human subjects treated with AAV2 or AAV1 against the AAV capsid ( Manno et al Nat Med 2006 ; Mingozzi et al Nat Med 2007 ; Brantly et al PNAS 2009 ; Mingozzi et al blood 2009 ), long term expression of the therapeutic gene is possible depending on the target tissue and the route of administration ( Brantly et al PNAS 2009 ; Simonelli et al mol therapy 2010 ).
  • AAV9 adeno-associated viruses of wide tropism
  • AAV9 viruses have shown efficient transduction in a broad range of tissues, with high tropism for liver, heart and skeletal muscle ( Inagaki et al Molecular Therapy 2006 ) and thus the beneficial effects of gene therapy can be achieved in more tissues.
  • AAV9 vectors have the unique ability to cross the blood-brain-barrier and target the brain upon intravenous injection in adult mice and cats ( Foust et al Nature biotechnology 2009 ; Duque et al Molecular therapy et al 2009 ).
  • the capsid (which is the part of the virus which determines the virus tropism) of the adeno-associated virus- based vector is made of capsid proteins of the serotype 9 adeno-associated virus (AAV9).
  • the polynucleotide sequence packed in the capsid is flanked by internal terminal repeats (ITRs) of an adeno-associated virus, preferably of serotype 2 which has been extensively characterised in the art, and presents a coding sequence located between the ITRs.
  • the nucleic acid preferably codes for a functional TERT polypeptide.
  • the regulatory sequence operatively linked to the TERT coding sequence is the cytomegalovirus promoter (CMV), although other suitable regulatory sequences will be known to those of skill in the art.
  • CMV cytomegalovirus promoter
  • AAV serotype for the capsid protein of the gene therapy vector may be thus based on the desired site of gene therapy. If the target tissue is skeletal muscle, for example, in treating loss of neuromuscular coordination, AAV1- and AAV6-based viral vectors can be used. Both of these serotypes are more efficient at trans fecting muscle than other AAV serotypes. AAV3 is useful for trans fecting haematopoietic cells.
  • viral vectors can be used in the present invention.
  • Any vector compatible with use in gene therapy can be used in the present invention.
  • Heilbronn & Weger (2010) Handb Exp Pharmacol. 197: 143-70 provides a review of viral vectors that are useful in gene therapy.
  • vectors comprising a coding sequence for telomerase reverse transcriptase (TERT) suitable for use in gene therapy are an important point for putting the invention into practice.
  • Suitable gene therapy vectors include any kind of particle that comprises a polynucleotide fragment encoding the telomerase reverse transcriptase (TERT) protein, operably linked to a regulatory element such as a promoter, which allows the expression of a functional TERT protein demonstrating telomerase reverse transcriptase activity in the targeted cells.
  • TERT is encoded by the nucleic acid sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 3, or is an active fragment or functional equivalent of TERT.
  • gene therapy vector includes within its scope naked DNA molecules such as plasmids or mini-circles, i.e. circular DNA molecules which do not contain bacterial DNA sequences, provided that the TERT coding sequence and its linked regulatory element are inserted in the plasmid, as well as to more complicated systems, such as particles with the structure of virions (viral particles), comprising at least a capsid and at least a polynucleotide sequence, with a size that allows the polynucleotide sequence to be packed within the capsid in a manner similar to that of the native genome of the virus of origin of the capsid.
  • naked DNA molecules such as plasmids or mini-circles, i.e. circular DNA molecules which do not contain bacterial DNA sequences, provided that the TERT coding sequence and its linked regulatory element are inserted in the plasmid, as well as to more complicated systems, such as particles with the structure of virions (viral particles), comprising at least a capsid and at least a polynu
  • the polynucleotide sequence must include a region where the TERT coding sequence and its linked regulatory element are inserted such that the telomerase reverse transcriptase protein can be expressed from that polynucleotide sequence once the viral particle has infected a cell.
  • the gene therapy vector suitable for being used in the invention is a non- integrative vector, such as an adeno-associated virus-based non-integrative vector.
  • a non-integrative vector such as an adeno-associated virus-based non-integrative vector.
  • non-integrative vectors incorporate a safety mechanism to avoid over-proliferation of TERT expressing cells that will lose the vector if the cells start proliferating quickly.
  • the regulatory sequence operatively linked to the TERT coding sequence is the cytomegalovirus promoter (CMV).
  • CMV cytomegalovirus promoter
  • the nucleic acid sequence encoding TERT is operably linked to a regulatory sequence that drives the expression of the coding sequence.
  • regulatory element means a nucleic acid sequence that serves as a promoter, i.e., regulates expression of a nucleic acid sequence operably linked to the promoter.
  • Such "regulatory elements” or “promoters” can control the expression of linked nucleic acid sequences either constitutively or inducible.
  • the regulatory sequence may be a constitutive promoter.
  • An example of a regulatory sequence which is a constitutive promoter is the cytomegalovirus (CMV) promoter.
  • TERT expression persists for a time of several months to several years. In mice, TERT expression was detectable after 5 months. In monkey, gene expression following gene therapy with an AAV- based vector has been detected up to 6 years after treatment and up to 8 years in dogs ( Rivera et al Blood 2005 , and Niemeyer et al blood 2009 ). Frequent repetition of treatment using the methods and vectors of the invention is therefore not necessary.
  • the subject is treated once. In an alternative embodiment, the subject is treated initially, and is then treated again once TERT expression levels decrease by about 50% of those attained immediately following treatment.
  • Treatment may be repeated with the same or alternative vector to maintain the reduction in age-related disorders if necessary, for example annually, or once every 5 years or once a decade.
  • the second and subsequent administrations may be a vector with a capsid derived from a different serotype than that used for the first administration. It is possible that a subject may develop neutralising antibodies to the first gene therapy vector, making it ineffective if administered a second or subsequent time ( Amado et al (2010) Science Translational Medicine 2(21):21ral6 ).
  • the methods of treatment of the invention have the effect of treating and/or preventing conditions associated with short telomere length.
  • the invention refers to a gene therapy method or the use of a nucleic acid vector as described above, for use in the treatment or prevention in a subject of condition associated with short telomere length, including but not limited to genetically based conditions such as Dyskeratosis congenita, Aplastic anaemia, Myelodysplastic Syndrome, Fanconi anaemia, and pulmonary fibrosis.
  • the effectiveness of treatment of the conditions associated with short telomere length can be measured by various methods known in the art.
  • the effectiveness of the treatment is measured by an increase in lifespan of a treated patient suffering from a condition associated with short telomere length as compared to the expected lifespan of an untreated patient suffering from the same condition.
  • the lifespan is extended by 5%, 10%, 15%, 20% or more, with reference to the expected lifespan for a patient suffering from the same condition.
  • the effectiveness of the treatment is measured by a delayed or prevented bone marrow failure in a treated patient suffering from a condition associated with short telomere as compared to the expected onset of bone marrow failure in an untreated patient suffering from the same condition.
  • the delay in the onset of bone marrow failure of a treated patient suffering from a condition associated with short telomere length is extended by 5%, 10%, 15%, 20% or more, with reference to the expected onset of bone marrow failure for an untreated patient suffering from the same condition.
  • the effectiveness of the treatment is measured by an increase in overall fitness of a treated patient suffering from a condition associated with short telomere length treated as compared to the overall fitness of an untreated patient suffering from the same condition.
  • Overall fitness can be determined by measuring physical attributes associated with the particular condition. Examples of such physical attributes include skin abnormalities (such as skin hyperpigmentation), premature aging (such as hair greying, nail dystrophy, oral leucoplakia), and anaemic pallor. Dokal, I. 2011. Hematology Am Soc Hematol Educ Program, 480-486 .
  • an increase in overal fitness can be determined by a decrease in physical attributes associated with the particular condition exhibited by the treated patient.
  • Overall fitness can also be measure by determining the blood count of the patient.
  • increased overall fitness is measured by determining the amount of leukocytes, lymphocytes, thrombocytes in a peripheral blood sample. Higher blood count indicates an increased overall fitness. In certain embodiments, the blood count in a treated patient is increased by 5%, 10%, 15%, 20% or more, with reference to the blood count of an untreated patient suffering from the same condition.
  • the efficacy of the treatment can also be measured by directly determining telomere length in sample taken from the patient.
  • Telomere length can be measured, for example, by using standard hybridization techniques, such as fluorescence in situ hybridization (FISH), Quantitative Fluorescent in situ hybridization (Q-FISH), or High Throughput Quantitative Fluorescent in situ hybridization (HT Q-FISH). (Gonzalez-Suarez, Samper et al. 2001)in a sample taken from the patient, Samples suitable for telomere analysis include bone marrow tissue and blood samples. Telomere length can also be measured as described in Slagboom et al or Canela et al. (2007, PNAS 104:5300-5305 ).
  • samples are taken from the patient undergoing treatment throughout the course of the treatment so that both absolute telomere length and the rate of telomere shortening over the course of treatment can be determined. Samples may be taken every day during the course of treatment, or at longer intervals. In one embodiment, samples are taken once a week, once every two week, once every three weeks, once every 4 weeks, once every five weeks, once every six weeks or longer. Comparison of telomere length can be measured by a comparing the proportion of short telomeres in a sample taken from a patient.
  • the proportion of short telomeres is the fraction of telomeres presenting an intensity below the mean intensity of the sample as measured by a in situ hybridization technique, such as FISH or Q-FISH.
  • the proportion of short telomeres is the fraction of telomeres presenting an intensity 75%, 70%, 65%, 60%, 55%, 50%, 45%, 40% or more below the mean intensity of the sample.
  • the proportion of short telomeres is the fraction of telomeres presenting an intensity 50% or more below the mean intensity of the sample.
  • the proportion of short telomeres is the fraction of telomeres below a certain length, e.g. 8 kb, 7 kb, 6 kb, 5 kb, or shorter In one embodiment, the proportion of short telomeres is the fraction of telomeres 8 kb or shorter . In another embodiment, the proportion of short telomeres is the fraction of telomeres 7 kb or shorter. In another embodiment, the proportion of short telomeres is the fraction of telomeres 6 kb or shorter. In another embodiment, the proportion of short telomeres is the fraction of telomeres 5 kb or shorter. In another embodiment, the proportion of short telomeres is the fraction of telomeres 4 kb or shorter. In another embodiment, the proportion of short telomeres is the fraction of telomeres 3 kb or shorter.
  • the effectiveness of the treatment is measured by a decrease in the proportion of short telomeres in sample taken from a treated patient suffering from a condition associated with short telomere length as compared to a control sample.
  • the proportion of short telomeres in a sample taken from a treated patient is decreased by 10%, 20%, 30%, 40%, 50%, 60%, 70%, or greater as compared to a control sample.
  • the control sample is a sample taken from the same patient prior to the treatment, or taken at an earlier stage of the treatment.
  • the control sample is a sample taken from a patient suffering from the same condition and not provided the treatment.
  • the invention is applied to the subject by administering a pharmaceutical composition comprising an effective amount of any one of the gene therapy vectors compatible with the invention described above.
  • a “pharmaceutical composition” is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vitro, in vivo or ex vivo.
  • Composition is intended to mean a combination of active agent and another compound or composition, inert (for example, a detectable agent or label) or active.
  • An “effective amount” is an amount sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages.
  • compositions will generally be administered to a subject in aqueous form. Prior to administration, however, the composition may have been in a non-aqueous form. For instance, although some viral vectors are manufactured in aqueous form, then filled and distributed and administered also in aqueous form, other viral vectors are lyophilised during manufacture and are reconstituted into an aqueous form at the time of use. Thus a composition of the invention may be dried, such as a lyophilised formulation.
  • the composition may include preservatives such as thiomersal or 2-phenoxyethanol. It is preferred, however, that the composition should be substantially free from (i.e. less than 5 ⁇ g/ml) mercurial material e.g. thiomersal-free.
  • a physiological salt such as a sodium salt.
  • Sodium chloride (NaCl) is preferred, which may be present at between 1 and 20 mg/ml e.g. about 10+2mg/ml NaCl.
  • Other salts that may be present include potassium chloride, potassium dihydrogen phosphate, disodium phosphate dehydrate, magnesium chloride, calcium chloride, etc.
  • Compositions will generally have an osmolality of between 200 mOsm/kg and 400 mOsm/kg, preferably between 240-360 mOsm/kg, and will more preferably fall within the range of 290-310 mOsm/kg.
  • Compositions may include one or more buffers.
  • Typical buffers include: a phosphate buffer; a Tris buffer; a borate buffer; a succinate buffer; a histidine buffer (particularly with an aluminum hydroxide adjuvant); or a citrate buffer. Buffers will typically be included in the 5-20mM range.
  • the composition may include material for a single administration, or may include material for multiple administrations (i.e. a 'multidose' kit).
  • a preservative is preferred in multidose arrangements.
  • the compositions may be contained in a container having an aseptic adaptor for removal of material.
  • compositions of the invention for use in humans are typically administered in a dosage volume of about 0.5ml, although a half dose (i.e. about 0.25ml) may be administered to children.
  • the invention also provides a nucleic acid sequence encoding a TERT for use in therapy.
  • the invention also provides a nucleic acid vector comprising a coding sequence for telomerase reverse transcriptase (TERT), for use in a method of therapy and a gene therapy vector comprising a coding sequence for telomerase reverse transcriptase (TERT), for use in a method of therapy.
  • the therapy may be treating or preventing a condition associated with short telomere length.
  • the TERT nucleic acid sequence may be the sequence as recited in SEQ ID NO: 1 or SEQ ID NO: 3 or a fragment or functional equivalent thereof.
  • the TERT protein may have a sequence as recited in SEQ ID NO: 2 or SEQ ID NO: 4, or a fragment or functional equivalent thereof.
  • patient refers to a mammal.
  • the patient is a rodent, primate, ungulate, cat, dog, or other domestic pet or domesticated mammal.
  • the mammal is a mouse, rat, rabbit, pig, horse, sheep, cow, domestic cat or dog, or a human.
  • the patient is a human.
  • mice of C57B6 background that are homozygous carrier of a conditional TRF1 transgene (TRF1 flox/flox ) and further are transgenic for the Cre-recombinase under the control of the endogenous and interferon-inducible Mx1 promoter will be used to test the efficacy of the telomerase gene therapy to treat Dyskeratosis congenital (DKC).
  • DKC Dyskeratosis congenital
  • mice One month after transplantation the mice will be injected via their tail vein with 4 ⁇ 10 12 AAV9 genomes carrying the mTERT cDNA under the control of the potent cytomegalovirus promoter (for virus production, see below 3.3).
  • AAV9 lacking the telomerase gene will be injected into a control group.
  • a separate group of animals will be injected with AAV9-eGFP.
  • TRF1 deletion in the bone marrow will be induced by long-term polyinosinic-polycytidylic acid (pI:pC) treatment with intraperitoneal injections every third day.
  • pI:pC acts as immunostimulant and activates Cre expression which in turn leads to TRF1 deletion in approximately 50% of hematopoietic cells (upon each injection) with the above mentioned consequences (see 2).
  • pI:pC treatment animal groups previously infected with AAV9-mTERT and AAV9-empty will not undergo pI:pC treatment to serve as additional control cohorts.
  • telomere shortening owed to compensatory proliferation in the remaining hematopoietic cells which have not lost TRF1, by ectopic expression of telomerase, will be reduced.
  • end point of experiment death of animals.
  • successful telomerase treatment should show improvements with regards to overall fitness of the animals, i.e. no skin abnormalities and no anaemic pallor. The latter goes hand in hand with higher blood counts (leukocytes, lymphocytes, thrombocytes), which will be determined from peripheral blood samples.
  • the efficacy of telomerase expression on the molecular level will include telomere length measurements.
  • AAV based viral vectors for transduction will be generated by triple transfection of HEK293T cells as described in (Matsushita et al., 1998). Briefly, to 80% confluence grown cells are co-transfected with plasmids (1) carrying the expression cassette flanked by the AAV9 viral ITRs, (2) a helper plasmid carrying the AAV rep2 and cap9 genes, and (3) a plasmid carrying the adenovirus helper functions.
  • the expression cassettes harbour murine TERT under the control of CMV promoter plus 3'-UTR (AAV9-mTERT), CMV promoter (AAV9-empty) alone and eGFP under the control of CMV promoter and SV40 polyA signal (AAV9-eGFP).
  • Vectors are purified following an optimised method based on two consecutive cesium chloride gradients (Ayuso et al., 2010). Titres of viral genomes particles are determined by quantitative real time PCR. Viruses can be stably kept at -80°C until infection of animals.
  • RNA from tissues is extracted with Trizol (Life Technologies). RNA samples are DNase I treated and are used as template for a reverse transcription reaction using random primers and Superscript Reverse Transcriptase (Life Technologies), according to the manufacturer's guidelines. Quantitative real-time PCR is performed using an ABI PRISM 7700 (Applied Biosystems), using DNA Master SYBR Green I mix (Applied Biosystems).
  • the primers are the primers:
  • Trf1 lox / lox Mx1-Cre and Trf1 lox / lox Mx1-wt mice were generated previously described (Martinez et al., 2009) ENREF 20 .
  • Trf1 lox / lox Mx1-Cre mice were used as bone marrow donors for transplantation into 8 weeks old lethally (12Gy) irradiated wild-type mice as previously described (Beier et al., 2012, Samper et al., 2002).
  • mice were intraperitoneally injected with polyinosinic-polycytidylic acid (pI:pC; Sigma-Aldrich) (15 ug/g body weight) 3 times per week for a total duration of 5 weeks. Mice were left for an additional week before they were randomly assigned to two groups for the treatment with AAV9 -Tert or AAV9-empty gene therapy vectors. Vectors were administered via tail vein injection at a concentration of 4 ⁇ 10E12 viral genomes per mouse.
  • Viral vectors were generated as described previously (Matsushita et al., 1998) and purified described in (Ayuso et al., 2010). Briefly, vectors were produced through triple transfection of HEK293T. Cells were grown in roller bottles (Corning, NY, USA) in Dulbecco's Modified Eagle's Medium supplemented with FBS (10% v/v) to 80% confluence and then co-transfected with: plasmid-1 carrying the expression cassette for gene of interest flanked by the AAV2 viral ITRs; plasmid-2 carrying the AAV rep2 and cap9 genes; plasmid-3 carrying the adenovirus helper functions (plasmids were kindly provided by K.A.
  • CMV cytomegalovirus
  • AAV9 particles were purified following an optimized method using two caesium chloride gradients, dialysed against PBS, filtered and stored at -80°C until use (Ayuso et al., 2010).
  • Viral genomes particles titres were determined by a standardized quantitative real time PCR method(Ayuso et al., 2014) and primers specific for the CMV sequence:
  • Bone marrow samples (sternum or tibia bone) were fixed in phosphate-buffered 4% formaldehyde and bones after decalcification paraffin embedded. 5 ⁇ m tissue sections were stained with Hematoxylin-Eosin for histological bone marrow assessment. Immunohistochemistry was performed on deparaffinized tissues sections. After antigen retrieval samples were processed with the anti-EGFP antibodies (rabbit anti-EGFP, 1:200; Abcam, ab290). EGFP positive cells were counted in a semi automated way using ImageJ software.
  • Cells were then resuspended in FACS buffer at 20-25 ⁇ 10 ⁇ 6 cell / ml and the antibody cocktail was added as follows: Anti-sca-1-PerCP-Cy5.5 (1:200), lin cocktail-eFluor450 (1:50) (all eBioscience), and anti-c-kit-APC-H7 (1:100) (BD Pharmingen). Cells were incubated for 30 min.
  • DAPI 200 g/mL was added and cells were subsequently sorted in a FACS ARIA IIu (Becton Dickinson, San Jose, CA) into HSCs (lin negatice, sca1 and c-kit positive) and lineage positive (lin positive) fractions.
  • Short-term colony-forming assay was performed by plating 1 ⁇ 10 4 and 2 ⁇ 10 4 freshly isolated mononucleated bone marrow cells (erythrocytes were lysed as described above) in 35-mm dishes (StemCell Technologies) containing Methocult (methylcellulose-based) media (StemCell Technologies) as described in the manufacturer's protocol. All experiments were performed in duplicates and the numbers of colonies formed were counted after 12 days incubation at 37 °C.
  • Peripheral blood was drawn from the facial vein ( ⁇ 50 ⁇ l) and collected into anti-coagulation tubes (EDTA). Blood counts were determined using an Abacus Junior Vet veterinary hematology analyzer.
  • RNA from whole bone marrow extracts or FACS sorted bone marrow cells wass isolated using Qiagen's RNeasy mini kit according to the manufacturer protocol. The optional DNaseI digest was always performed. Quantitative real-time PCR was performed using an ABI PRISM 7700 or QuantStudio 6 Flex (both Applied Biosystems). Primers sequences for Tert and reference genes Act1 and TBP are as follows:
  • telomere probe mix 10 min air-dried and 30 ⁇ l of telomere probe mix added to each slide (10mM TrisCl pH 7, 25mM MgCl2, 9mM citric acid, 82mM Na2HPO4, 70% deionized formamide (Sigma), 0.25% blocking reagent (Roche) and 0.5 mg/ml Telomeric PNA probe (Panagene)), a cover slip added and slides incubated for 3 min at 85 °C, and 2 h at room temperature in a wet chamber in the dark.
  • HT-Q-FISH on peripheral blood leukocytes was done as described (Canela et al., 2007a). Briefly, 120-150 ⁇ l of blood were extracted from the facial vein. Erythrocytes were lysed (Erythrocyte lysis buffer, Qiagen) and 30-90 k leukocytes were plated in duplicate into clear-bottom, black-walled 96-well plates pre-coated for 30 min with 0.001% poly-L-lysine. Plates were incubated at 37°C for 2 h and fixed with methanol/acetic acid (3:1, v/v) 2 ⁇ 10 min and overnight at -20°C.
  • telomere-specific PNA-CY3 probe was used to stain nuclei.
  • TD values were analysed using individual telomere spots (>10,000 telomere spots per sample). The average fluorescence intensities of each sample were converted into kilobase using L5178-R and L5178-S cells as calibration standards, which have stable TLs of 79.7 and 10.2 kb, respectively. Samples were analyzed in duplicate.
  • AAV9-Tert targets bone marrow and hematopoietic stem cells
  • AAV9 vectors to transduce the bone marrow upon intravenous injection by using both a AAV9-EGFP reporter virus, which allows determination of the location and percentage of AAV9-transduced cells, as well as by determining Tert mRNA expression in vivo in different bone marrow cell populations following AAV9-Tert treatment.
  • AAV9-EGFP reporter virus allows determination of the location and percentage of AAV9-transduced cells, as well as by determining Tert mRNA expression in vivo in different bone marrow cell populations following AAV9-Tert treatment.
  • Trf1 deletion can be induced by administration of pl:pC and subsequent expression of the Cre recombinase (Beier et al., 2012).
  • Trf1 deletion by injecting mice 3 times per week with pI:pC for a total period of 5 weeks, at which point these mice start showing signs of aplastic anemia (Beier et al., 2012).
  • mice were subjected to gene therapy treatment with AAV9 -Tert or AAV9-empty control vectors.
  • mice Post mortem histopathologic analysis of bone marrow sections from mice that died during the first 100 days further confirmed the aplastic anemia phenotype.
  • mice presented with severe bone marrow hypo- and aplasia in 2 or all 3 blood lineages. While the diagnosis at the point of death in both groups was marrow bone failure and aplasia the phenotype appeared milder in the AAV9-Tert group compared with the AAV9-empty group as seen by higher bone marrow cellularity.
  • telomere treatment leads to telomere elongation in peripheral blood and bone marrow
  • telomere length in mice treated with telomerase was compared to mice receiving the control vector.
  • HT-Q-FISH technology Canela et al., 2007b
  • telomere length in peripheral blood monocytes was extracted blood at 4 different time points; after bone marrow engraftment (1), after pI:pC treatment (2), 2 months after AAV9 injection (3) and 4 months after AAV9 injection (4).
  • telomere length between time point 1 and 2 in both groups drops by approximately 10kb which is owed to the pI:pC treatment.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Genetics & Genomics (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Environmental Sciences (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Biomedical Technology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pulmonology (AREA)
  • Hematology (AREA)
  • Diabetes (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Enzymes And Modification Thereof (AREA)
EP21198809.2A 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase Pending EP3978031A1 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP14382312 2014-08-08
PCT/EP2015/067874 WO2016020345A1 (fr) 2014-08-08 2015-08-04 Thérapies à base de la transcriptase inverse télomoérase
EP21151155.5A EP3848056B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP18215767.7A EP3485914B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP15744254.2A EP3193943B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de la transcriptase inverse télomoérase

Related Parent Applications (4)

Application Number Title Priority Date Filing Date
EP18215767.7A Division EP3485914B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP15744254.2A Division EP3193943B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de la transcriptase inverse télomoérase
EP21151155.5A Division EP3848056B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP21151155.5A Division-Into EP3848056B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase

Publications (1)

Publication Number Publication Date
EP3978031A1 true EP3978031A1 (fr) 2022-04-06

Family

ID=51454634

Family Applications (4)

Application Number Title Priority Date Filing Date
EP18215767.7A Active EP3485914B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP21198809.2A Pending EP3978031A1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP21151155.5A Active EP3848056B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP15744254.2A Active EP3193943B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de la transcriptase inverse télomoérase

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP18215767.7A Active EP3485914B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase

Family Applications After (2)

Application Number Title Priority Date Filing Date
EP21151155.5A Active EP3848056B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de transcriptase inverses de la télomérase
EP15744254.2A Active EP3193943B1 (fr) 2014-08-08 2015-08-04 Thérapies à base de la transcriptase inverse télomoérase

Country Status (13)

Country Link
US (3) US11529396B2 (fr)
EP (4) EP3485914B1 (fr)
JP (1) JP6676262B2 (fr)
CY (1) CY1124012T1 (fr)
DK (2) DK3193943T3 (fr)
ES (2) ES2864731T3 (fr)
HR (1) HRP20210265T1 (fr)
HU (1) HUE053526T2 (fr)
LT (1) LT3485914T (fr)
PL (1) PL3485914T3 (fr)
PT (1) PT3485914T (fr)
SI (1) SI3485914T1 (fr)
WO (1) WO2016020345A1 (fr)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112063601B (zh) * 2020-09-22 2023-06-02 浙江愈方生物科技有限公司 一种灭活性端粒酶、具有其的腺病毒和人造mRNA及应用
EP4089171A1 (fr) 2021-05-12 2022-11-16 Fundación del Sector Público Estatal Centro Nacional de Investigaciones Oncológicas Carlos III (F.S.P. CNIO) Génomes et vecteurs viraux recombinants à encodage tert
WO2023138791A1 (fr) * 2022-01-24 2023-07-27 Fundación Del Sector Público Estatal Centro Nacional De Investigaciones Oncológicas Carlos III (F.S.P. CNIO) Thérapie par transcriptase inverse de la télomérase de la fibrose rénale et animaux non humains associés

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010018731A2 (fr) * 2008-08-12 2010-02-18 Japan Health Sciences Foundation Arn polymérase dépendante de l'arn mammalienne
EP2402038A1 (fr) * 2010-07-02 2012-01-04 Fundación Centro Nacional De Investigaciones Oncológicas Carlos III Transcriptase inverse de la télomérase pour la protection contre le vieillissement

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7569385B2 (en) * 2003-08-14 2009-08-04 The Regents Of The University Of California Multipotent amniotic fetal stem cells
US20110151447A1 (en) * 2007-11-06 2011-06-23 Children's Medical Center Corporation Method to produce induced pluripotent stem (ips) cells from non-embryonic human cells
WO2014105870A1 (fr) * 2012-12-27 2014-07-03 Sierra Sciences, Inc. Amélioration de la santé chez des mammifères par une thérapie génique impliquant la transcriptase inverse de la télomérase

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010018731A2 (fr) * 2008-08-12 2010-02-18 Japan Health Sciences Foundation Arn polymérase dépendante de l'arn mammalienne
EP2402038A1 (fr) * 2010-07-02 2012-01-04 Fundación Centro Nacional De Investigaciones Oncológicas Carlos III Transcriptase inverse de la télomérase pour la protection contre le vieillissement

Non-Patent Citations (102)

* Cited by examiner, † Cited by third party
Title
ALTSCHUL SFGISH WMILLER WMYERS EWLIPMAN DJ: "Basic local alignment search tool", J MOL BIOL, vol. 215, no. 3, 1990, pages 403 - 410, XP002949123, DOI: 10.1006/jmbi.1990.9999
AMADO ET AL., SCIENCE TRANSLATIONAL MEDICINE, vol. 2, no. 21, 2010, pages 21 - 16
ARMANIOS, M.: "An emerging role for the conserved telomere component 1 (CTC1) in human genetic disease", PEDIATR BLOOD CANCER, vol. 59, 2012, pages 209 - 210
ARMANIOS, M.BLACKBURN, E.H.: "The telomere syndromes. Nature reviews", GENETICS, vol. 13, 2012, pages 693 - 704
AYUSO, E.F. MINGOZZIJ. MONTANEX. LEONX. M. ANGUELAV. HAURIGOTS. A. EDMONSONL. AFRICAS. ZHOUK. A. HIGH: "High AAV vector purity results in serotype- and tissue-independent enhancement of transduction efficiency", GENE THER, vol. 17, 2010, pages 503 - 510, XP055073587, DOI: 10.1038/gt.2009.157
AYUSO, E.MINGOZZI, F.MONTANE, J.LEON, X.ANGUELA, X.M.HAURIGOT, V.EDMONSON, S.A.AFRICA, L.ZHOU, S.HIGH, K.A. ET AL.: "High AAV vector purity results in serotype- and tissue-independent enhancement of transduction efficiency", GENE THERAPY, vol. 17, 2010, pages 503 - 510, XP055073587, DOI: 10.1038/gt.2009.157
AYUSO, E.V. BLOUINM. LOCKS. MCGORRAYX. LEONM. R. ALVIRAA. AURICCHIOS. BUCHERA. CHTARTOK. R. CLARK: "Manufacturing and Characterization of a Recombinant Adeno-Associated Virus Type 8 Reference Standard Material.", HUM GENE THER., 2014
BALL, S. E.F. M. GIBSONS. RIZZOJ. A. TOOZEJ. C. MARSHE. C. GORDON-SMITH: "Progressive telomere shortening in aplastic anemia", BLOOD, vol. 91, 1998, pages 3582 - 3592
BEIER, F.FORONDA, M.MARTINEZ, P.BLASCO, M.A.: "Conditional TRF1 knockout in the hematopoietic compartment leads to bone marrow failure and recapitulates clinical features of Dyskeratosis congenita", BLOOD, 2012
BERNARDES DE JESUS DE JESUS, B.BLASCO, M.A: "Telomerase at the intersection of cancer and aging", TRENDS GENET, vol. 29, 2013, pages 513 - 520
BERNARDES DE JESUS, B.E. VERAK. SCHNEEBERGERA. M. TEJERAE. AYUSOF. BOSCHM. A. BLASCO: "Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer", EMBO MOL MED, vol. 4, 2012, pages 1 - 14
BERNARDES DE JESUS, B.VERA, E.SCHNEEBERGER, K.TEJERA, A.M.AYUSO, E.BOSCH, F.BLASCO, M.A.: "Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer", EMBO MOLECULAR MEDICINE, vol. 4, 2012, pages 691 - 704, XP055481728, DOI: 10.1002/emmm.201200245
BLACKBURN, E. H.: "Switching and signaling at the telomere", CELL, vol. 106, 2001, pages 661 - 673, XP002261021, DOI: 10.1016/S0092-8674(01)00492-5
BLASCO, M.A.: "Telomere length, stem cells and aging", NATURE CHEMICAL BIOLOGY, vol. 3, 2007, pages 640 - 649
BLASCO, M.A.LEE, H.W.HANDE, M.P.SAMPER, E.LANSDORP, P.M.DEPINHO, R.A.GREIDER, C.W.: "Telomere shortening and tumor formation by mouse cells lacking telomerase RNA", CELL, vol. 91, 1997, pages 25 - 34
BLASCO, NAT CHEM BIOL, vol. 3, 2007, pages 640 - 649
BRANTLY ET AL., PNAS, 2009
BRUNO BERNARDES DE JESUS ET AL: "Telomerase gene therapy in adult and old mice delays aging and increases longevity without increasing cancer : TERT alone extends lifespan of adult/old mice", EMBO MOLECULAR MEDICINE, vol. 4, no. 8, 15 May 2012 (2012-05-15), US, pages 691 - 704, XP055481728, ISSN: 1757-4676, DOI: 10.1002/emmm.201200245 *
BUNING, H.PERABO, L.COUTELLE, O.QUADT-HUMME, S.HALLEK, M.: "Recent developments in adeno-associated virus vector technology", THE JOURNAL OF GENE MEDICINE, vol. 10, 2008, pages 717 - 733
CALADO, R.T.YEWDELL, W.T.WILKERSON, K.L.REGAL, J.A.KAJIGAYA, S.STRATAKIS, C.A.YOUNG, N.S.: "Sex hormones, acting on the TERT gene, increase telomerase activity in human primary hematopoietic cells", BLOOD, vol. 114, 2009, pages 2236 - 2243
CALLEN, E.SAMPER, E.RAMIREZ, M.J.CREUS, A.MARCOS, R.ORTEGA, J.J.OLIVE, T.BADELL, I.BLASCO, M.A.SURRALLES, J.: "Breaks at telomeres and TRF2-independent end fusions in Fanconi anemia", HUM MOL GENET, vol. 11, 2002, pages 439 - 444
CANELA ET AL., PNAS, vol. 104, 2007, pages 5300 - 5305
CANELA, A.E. VERAP. KLATTM. A. BLASCO: "High-throughput telomere length quantification by FISH and its application to human population studies", PROC NATL ACAD SCI U S A, vol. 104, 2007, pages 5300 - 5305, XP055323118, DOI: 10.1073/pnas.0609367104
CANELA, A.P. KLATTM. A. BLASCO: "Telomere length analysis", METHODS MOL BIOL, vol. 371, 2007, pages 45 - 72
CARROLL, K.A.LY, H.: "Telomere dysfunction in human diseases: the long and short of it!", INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY, vol. 2, 2009, pages 528 - 543
DE LANGE, T.: "Shelterin: the protein complex that shapes and safeguards human telomeres", GENES DEV, vol. 19, 2005, pages 2100 - 2110
DOKAL, I.: "Dyskeratosis congenita", HEMATOLOGY AM SOC HEMATOL EDUC PROGRAM, 2011, pages 480 - 486
DOKAL, I.: "Dyskeratosis congenita. Hematology / the Education Program of the American Society of Hematology", AMERICAN SOCIETY OF HEMATOLOGY, 2011, pages 480 - 486
DOKAL, I.VULLIAMY, T.: "Inherited bone marrow failure syndromes", HAEMATOLOGICA, vol. 95, 2010, pages 1236 - 1240
DUQUE ET AL., MOLECULAR THERAPY, 2009
DUQUE, S.JOUSSEMET, B.RIVIERE, C.MARAIS, T.DUBREIL, L.DOUAR, A.M.FYFE, J.MOULLIER, P.COLLE, M.A.BARKATS, M.: "Intravenous administration of self-complementary AAV9 enables transgene delivery to adult motor neurons", MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 17, 2009, pages 1187 - 1196, XP055103985, DOI: 10.1038/mt.2009.71
ERIK R WESTIN ET AL: "Telomere restoration and extension of proliferative lifespan in dyskeratosis congenita fibroblasts", AGING CELL, BLACKWELL PUBLISHING, GB, vol. 6, no. 3, 1 June 2007 (2007-06-01), pages 383 - 394, XP008133670, ISSN: 1474-9718, [retrieved on 20070219], DOI: 10.1111/J.1474-9726.2007.00288.X *
FLORES ET AL., GENES AND DEV, vol. 22, 2008, pages 654 - 667
FLORES, I.A. CANELAE. VERAA. TEJERAG. COTSARELISM. A. BLASCO: "The longest telomeres: a general signature of adult stem cell compartments", GENES DEV, vol. 22, 2008, pages 654 - 667, XP002518436, DOI: 10.1101/gad.451008
FLORES, IM. L. CAYUELAM. A. BLASCO: "Effects of telomerase and telomere length on epidermal stem cell behavior", SCIENCE, vol. 309, 2005, pages 1253 - 1256
FOUST ET AL., NATURE BIOTECHNOLOGY, 2009
FOUST, K.D.NURRE, E.MONTGOMERY, C.L.HERNANDEZ, A.CHAN, C.M.KASPAR, B.K.: "Intravascular AAV9 preferentially targets neonatal neurons and adult astrocytes", NAT BIOTECHNOL, vol. 27, 2009, pages 59 - 65, XP055023143, DOI: 10.1038/nbt.1515
GADALLA, S.M.CAWTHON, R.GIRI, N.ALTER, B.P.SAVAGE, S.A.: "Telomere length in blood, buccal cells, and fibroblasts from patients with inherited bone marrow failure syndromes", AGING (ALBANY NY, vol. 2, 2010, pages 867 - 874
GAO, ALVIRA ET AL., PNAS, 2002
GAO, G.P.ALVIRA, M.R.WANG, L.CALCEDO, R.JOHNSTON, J.WILSON, J.M.: "Novel adeno-associated viruses from rhesus monkeys as vectors for human gene therapy", PROC NATL ACAD SCI U S A, vol. 99, 2002, pages 11854 - 11859
GENNARO, REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY, 2000
GHOSH, YUE ET AL., MOLECULAR THERAPY, 2007
GONZALEZ-SUAREZ, E.SAMPER, E.RAMIREZ, A.FLORES, J.M.MARTIN-CABALLERO, J.JORCANO, J.L.BLASCO, M.A.: "Increased epidermal tumors and increased skin wound healing in transgenic mice overexpressing the catalytic subunit of telomerase, mTERT, in basal keratinocytes", EMBO J, vol. 20, 2001, pages 2619 - 2630, XP002400668, DOI: 10.1093/emboj/20.11.2619
GREIDERBLACKBURN, CELL, vol. 43, 1985, pages 405 - 413
HARLEY, C. B.A. B. FUTCHERC. W. GREIDER: "Telomeres shorten during ageing of human fibroblasts", NATURE, vol. 345, 1990, pages 458 - 460
HEILBRONNWEGER, HANDB EXP PHARMACOL, vol. 197, 2010, pages 143 - 70
HERRERA, E.SAMPER, E.MARTIN-CABALLERO, J.FLORES, J.M.LEE, H.W.BLASCO, M.A.: "Disease states associated with telomerase deficiency appear earlier in mice with short telomeres", EMBO J, vol. 18, 1999, pages 2950 - 2960
HIROKI YAMAGUCHI ET AL: "Mutations in TERT, the Gene for Telomerase Reverse Transcriptase, in Aplastic Anemia", NEW ENGLAND JOURNAL OF MEDICINE, vol. 352, no. 14, 7 April 2005 (2005-04-07), pages 1413 - 1424, XP055213032, ISSN: 0028-4793, DOI: 10.1056/NEJMoa042980 *
HIROTOSHI SAKAGUCHI ET AL: "Inherited bone marrow failure syndromes in 2012", INTERNATIONAL JOURNAL OF HEMATOLOGY, SPRINGER JAPAN, JAPAN, vol. 97, no. 1, 28 December 2012 (2012-12-28), pages 20 - 29, XP035169874, ISSN: 1865-3774, DOI: 10.1007/S12185-012-1249-9 *
HIYAMA, E.K. HIYAMA: "Telomere and telomerase in stem cells", BR J CANCER, vol. 96, 2007, pages 1020 - 1024, XP055061521, DOI: 10.1038/sj.bjc.6603671
HOLME, H.HOSSAIN, U.KIRWAN, M.WALNE, A.VULLIAMY, T.DOKAL, I.: "Marked genetic heterogeneity in familial myelodysplasia/acute myeloid leukaemia", BRITISH JOURNAL OF HAEMATOLOGY, vol. 158, 2012, pages 242 - 248
INAGAKI ET AL., MOLECULAR THERAPY, 2006
INAGAKI, K.FUESS, S.STORM, T.A.GIBSON, G.A.MCTIERNAN, C.F.KAY, M.A.NAKAI, H.: "Robust systemic transduction with AAV9 vectors in mice: efficient global cardiac gene transfer superior to that of AAV8. Molecular therapy", THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY, vol. 14, 2006, pages 45 - 53
JAIME-PEREZ, J.C.COLUNGA-PEDRAZA, P.R.GOMEZ-RAMIREZ, C.D.GUTIERREZ-AGUIRRE, C.H.CANTU-RODRIGUEZ, O.G.TARIN-ARZAGA, L.C.GOMEZ-ALMAG: "Danazol as first-line therapy for aplastic anemia", ANNALS OF HEMATOLOGY, vol. 90, 2011, pages 523 - 527, XP019893253, DOI: 10.1007/s00277-011-1163-x
JASKELIOFF, M.MULLER, F.L.PAIK, J.H.THOMAS, E.JIANG, S.ADAMS, A.C.SAHIN, E.KOST-ALIMOVA, M.PROTOPOPOV, A.CADINANOS, J. ET AL.: "Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice", NATURE, vol. 469, 2011, pages 102 - 106
JIANG, H.LILLICRAP, D.PATARROYO-WHITE, S.LIU, T.QIAN, X.SCALLAN, C.D.POWELL, S.KELLER, T.MCMURRAY, M.LABELLE, A. ET AL.: "Multiyear therapeutic benefit of AAV serotypes 2, 6, and 8 delivering factor VIII to hemophilia A mice and dogs", BLOOD, vol. 108, 2006, pages 107 - 115, XP055733373, DOI: 10.1182/blood-2005-
JIANG, LILLICRAP ET AL., BLOOD, 2006
KAPLITT, FEIGIN, LANCET, 2009
KAPLITT, M.G.: "Gene therapy clinical trials in the human brain. Protocol development and review of current applications", FRONTIERS OF NEUROLOGY AND NEUROSCIENCE, vol. 25, 2009, pages 180 - 188
KEE, Y.D'ANDREA, A.D.: "Molecular pathogenesis and clinical management of Fanconi anemia", J CLIN INVEST, vol. 122, 2012, pages 3799 - 3806, XP055267930, DOI: 10.1172/JCI58321
LEE, H.W.BLASCO, M.A.GOTTLIEB, G.J.HORNER, J.W., 2NDGREIDER, C.W.DEPINHO, R.A.: "Essential role of mouse telomerase in highly proliferative organs", NATURE, vol. 392, 1998, pages 569 - 574
LIPMAN, DJPEARSON, WR: "Rapid and sensitive protein similarity searches", SCIENCE, vol. 227, no. 4693, 1985, pages 1435 - 41, XP000941106, DOI: 10.1126/science.2983426
MACIEJEWSKI, J. P.S. ANDERSONP. KATEVASN. S. YOUNG: "Phenotypic and functional analysis of bone marrow progenitor cell compartment in bone marrow failure", BR J HAEMATOL, vol. 87, 1994, pages 227 - 234
MAGUIRE, A.M.SIMONELLI, F.PIERCE, E.A.PUGH, E.N., JR.MINGOZZI, F.BENNICELLI, J.BANFI, S.MARSHALL, K.A.TESTA, F.SURACE, E.M. ET AL.: "Safety and efficacy of gene transfer for Leber's congenital amaurosis", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 358, 2008, pages 2240 - 2248, XP055027817, DOI: 10.1056/NEJMoa0802315
MAGUIRE, SIMONELLI ET AL., NEJM, 2008
MANNO ET AL., NAT MED, 2006
MANNO ET AL., NAT MEDICINE, 2006
MANNO, C.S.PIERCE, G.F.ARRUDA, V.R.GLADER, B.RAGNI, M.RASKO, J.J.OZELO, M.C.HOOTS, K.BLATT, P.KONKLE, B. ET AL.: "Successful transduction of liver in hemophilia by AAV-Factor IX and limitations imposed by the host immune response", NATURE MEDICINE, vol. 12, 2006, pages 342 - 347, XP002578131
MARSH, J. C.S. E. BALLJ. CAVENAGHP. DARBYSHIREI. DOKALE. C. GORDON-SMITHJ. KEIDANA. LAURIEA. MARTINJ. MERCIECA: "Guidelines for the diagnosis and management of aplastic anaemia", BR J HAEMATOL, vol. 147, 2009, pages 43 - 70
MARTINEZ, P.BLASCO, M.A.: "Telomeric and extra-telomeric roles for telomerase and the telomere-binding proteins", NATURE REVIEWS. CANCER, vol. 11, 2011, pages 161 - 176, XP002696628, DOI: 10.1038/nrc3025
MARTINEZ, P.M. THANASOULAP. MUNOZC. LIAOA. TEJERAC. MCNEESJ. M. FLORESO. FERNANDEZ-CAPETILLOM. TARSOUNASM. A. BLASCO: "Increased telomere fragility and fusions resulting from TRF1 deficiency lead to degenerative pathologies and increased cancer in mice", GENES DEV, vol. 23, 2009, pages 2060 - 2075
MAS, A.MONTANE, J.ANGUELA, X.M.MUNOZ, S.DOUAR, A.M.RIU, E.OTAEGUI, P.BOSCH, F.: "Reversal of type 1 diabetes by engineering a glucose sensor in skeletal muscle", DIABETES, vol. 55, 2006, pages 1546 - 1553, XP002659438, DOI: 10.2337/db05-1615
MASON, P.J.BESSLER, M.: "The genetics of dyskeratosis congenita", CANCER GENETICS, vol. 204, 2011, pages 635 - 645
MATSUSHITA, T.ELLIGER, S.ELLIGER, C.PODSAKOFF, G.VILLARREAL, L.KURTZMAN, G.J.IWAKI, Y.COLOSI, P.: "Adeno-associated virus vectors can be efficiently produced without helper virus", GENE THERAPY, vol. 5, 1998, pages 938 - 945, XP002098259, DOI: 10.1038/sj.gt.3300680
MATSUSHITA, T.S. ELLIGERC. ELLIGERG. PODSAKOFFL. VILLARREALG. J. KURTZMANY. IWAKIP. COLOSI: "Adeno-associated virus vectors can be efficiently produced without helper virus", GENE THER, vol. 5, 1998, pages 938 - 945, XP002098259, DOI: 10.1038/sj.gt.3300680
MAVILIO, F.: "Gene therapies need new development models", NATURE, vol. 490, 2012, pages 7
MINGOZZI ET AL., NAT MED, 2007
NAKAO, S: "Immune mechanism of aplastic anemia", INT J HEMATOL, vol. 66, 1997, pages 127 - 134
NEAL S YOUNG ET AL: "Current concepts in the pathophysiology and treatment of aplastic anemia", BLOOD, 15 October 2006 (2006-10-15), United States, pages 2509 - 2519, XP055213196, Retrieved from the Internet <URL:http://bloodjournal.hematologylibrary.org/cgi/content/abstract/108/8/2509> DOI: 10.1182/blood-2006-03-010777 *
NIEMEYER ET AL., BLOOD, 2009
NIEMEYER, G.P.HERZOG, R.W.MOUNT, J.ARRUDA, V.R.TILLSON, D.M.HATHCOCK, J.VAN GINKEL, F.W.HIGH, K.A.LOTHROP, C.D., JR.: "Long-term correction of inhibitor-prone hemophilia B dogs treated with liver-directed AAV2-mediated factor IX gene therapy", BLOOD, vol. 113, 2009, pages 797 - 806
O'REILLY, M.SHIPP, A.ROSENTHAL, E.JAMBOU, R.SHIH, T.MONTGOMERY, M.GARGIULO, L.PATTERSON, A.CORRIGAN-CURAY, J.: "NIH oversight of human gene transfer research involving retroviral, lentiviral, and adeno-associated virus vectors and the role of the NIH recombinant DNA advisory committee", METHODS IN ENZYMOLOGY, vol. 507, 2012, pages 313 - 335
RIVERA ET AL., BLOOD, 2005
SAMPER, E.F. A. GOYTISOLOP. SLIJEPCEVICP. P. VAN BUULM. A. BLASCO: "Mammalian Ku86 protein prevents telomeric fusions independently of the length of TTAGGG repeats and the G-strand overhang", EMBO REP, vol. 1, 2000, pages 244 - 252, XP002558916, DOI: 10.1093/embo-reports/kvd051
SAMPER, E.FLORES, J.M.BLASCO, M.A.: "Restoration of telomerase activity rescues chromosomal instability and premature aging in Terc-/- mice with short telomeres", EMBO REP, vol. 2, 2001, pages 800 - 807
SAMPER, E.P. FERNANDEZR. EGUIAL. MARTIN-RIVERAA. BERNADM. A. BLASCOM. ARACIL: "Long-term repopulating ability of telomerase-deficient murine hematopoietic stem cells", BLOOD, vol. 99, 2002, pages 2767 - 2775
SAVAGE, S.A.ALTER, B.P.: "The role of telomere biology in bone marrow failure and other disorders", MECHANISMS OF AGEING AND DEVELOPMENT, vol. 129, 2008, pages 35 - 47, XP022460484
SAVAGE, S.A.CALADO, R.T.XIN, Z.T.LY, H.YOUNG, N.S.CHANOCK, S.J.: "Genetic variation in telomeric repeat binding factors 1 and 2 in aplastic anemia", EXPERIMENTAL HEMATOLOGY, vol. 34, 2006, pages 664 - 671, XP025017477, DOI: 10.1016/j.exphem.2006.02.008
SCOPES, J.M. BAGNARAE. C. GORDON-SMITHS. E. BALLF. M. GIBSON: "Haemopoietic progenitor cells are reduced in aplastic anaemia", BR J HAEMATOL, vol. 86, 1994, pages 427 - 430
SHI ET AL.: "AAV-based targeting gene therapy", AM. J. IMMUNOL., vol. 4, 2008, pages 51 - 65, XP055140785, DOI: 10.3844/ajisp.2008.51.65
SIMONELLI ET AL., MOL THERAPY, 2010
STROES ET AL., ATVB, 2008
STROES, E.S.NIERMAN, M.C.MEULENBERG, J.J.FRANSSEN, R.TWISK, J.HENNY, C.P.MAAS, M.M.ZWINDERMAN, A.H.ROSS, C.ARONICA, E. ET AL.: "Intramuscular administration of AAV1- lipoprotein lipase S447X lowers triglycerides in lipoprotein lipase-deficient patients", ARTERIOSCLEROSIS, THROMBOSIS, AND VASCULAR BIOLOGY, vol. 28, 2008, pages 2303 - 2304, XP055005048, DOI: 10.1161/ATVBAHA.108.175620
TAFURO, AYUSO ET AL., CARDIOVASCULAR RESEARCH, 2009
TAFURO, S.AYUSO, E.ZACCHIGNA, S.ZENTILIN, L.MOIMAS, S.DORE, F.GIACCA, M.: "Inducible adeno-associated virus vectors promote functional angiogenesis in adult organisms via regulated vascular endothelial growth factor expression", CARDIOVASCULAR RESEARCH, vol. 83, 2009, pages 663 - 671
TOMAS-LOBA, A.FLORES, I.FERNANDEZ-MARCOS, P.J.CAYUELA, M.L.MARAVER, A.TEJERA, A.BORRAS, C.MATHEU, A.KLATT, P.FLORES, J.M. ET AL.: "Telomerase reverse transcriptase delays aging in cancer-resistant mice", CELL, vol. 135, 2008, pages 609 - 622, XP002532531, DOI: 10.1016/j.cell.208.09.034
VULLIAMY, T.A. MARRONEI. DOKALP. J. MASON: "Association between aplastic anaemia and mutations in telomerase RNA", LANCET, vol. 359, 2002, pages 2168 - 2170, XP004798280, DOI: 10.1016/S0140-6736(02)09087-6
WALNE, A.J.VULLIAMY, T.BESWICK, R.KIRWAN, M.DOKAL, I.: "TINF2 mutations result in very short telomeres: analysis of a large cohort of patients with dyskeratosis congenita and related bone marrow failure syndromes", BLOOD, vol. 112, 2008, pages 3594 - 3600
WYNN, R. F.M. A. CROSSC. HATTONA. M. WILLL. S. LASHFORDT. M. DEXTERN. G. TESTA: "Accelerated telomere shortening in young recipients of allogeneic bone-marrow transplants", LANCET, vol. 351, 1998, pages 178 - 181, XP004832643, DOI: 10.1016/S0140-6736(97)08256-1
YAMAGUCHI, H.BAERLOCHER, G.M.LANSDORP, P.M.CHANOCK, S.J.NUNEZ, O.SLOAND, E.YOUNG, N.S.: "Mutations of the human telomerase RNA gene (TERC) in aplastic anemia and myelodysplastic syndrome", BLOOD, vol. 102, 2003, pages 916 - 918
YAMAGUCHI, H.CALADO, R.T.LY, H.KAJIGAYA, S.BAERLOCHER, G.M.CHANOCK, S.J.LANSDORP, P.M.YOUNG, N.S.: "Mutations in TERT, the gene for telomerase reverse transcriptase, in aplastic anemia", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 352, 2005, pages 1413 - 1424, XP055213032, DOI: 10.1056/NEJMoa042980
ZIEGLER, P.SCHREZENMEIER, H.AKKAD, J.BRASSAT, U.VANKANN, L.PANSE, J.WILOP, S.BALABANOV, S.SCHWARZ, K.MARTENS, U.M.: "Telomere elongation and clinical response to androgen treatment in a patient with aplastic anemia and a heterozygous hTERT gene mutation", ANNALS OF HEMATOLOGY, vol. 91, 2012, pages 1115 - 1120, XP037143572, DOI: 10.1007/s00277-012-1454-x

Also Published As

Publication number Publication date
EP3193943A1 (fr) 2017-07-26
EP3848056A1 (fr) 2021-07-14
LT3485914T (lt) 2021-03-25
EP3485914B1 (fr) 2021-01-13
JP6676262B2 (ja) 2020-04-08
DK3485914T3 (da) 2021-01-25
ES2864731T3 (es) 2021-10-14
CY1124012T1 (el) 2022-05-27
WO2016020345A1 (fr) 2016-02-11
EP3193943B1 (fr) 2019-03-06
US20170232075A1 (en) 2017-08-17
US20240075109A1 (en) 2024-03-07
US11529396B2 (en) 2022-12-20
US20210260169A1 (en) 2021-08-26
HUE053526T2 (hu) 2021-07-28
EP3485914A1 (fr) 2019-05-22
DK3193943T3 (da) 2019-06-11
PL3485914T3 (pl) 2021-08-02
EP3848056B1 (fr) 2024-02-14
SI3485914T1 (sl) 2021-04-30
PT3485914T (pt) 2021-03-16
JP2017524369A (ja) 2017-08-31
HRP20210265T1 (hr) 2021-04-02
ES2729872T3 (es) 2019-11-06

Similar Documents

Publication Publication Date Title
US20240075109A1 (en) Telomerase reverse transcriptase-based therapies
JP2022184901A (ja) モジュラーAAV送達システムによるCRISPR-Casゲノム編集
JP2017529854A (ja) 高効率ゲノム編集のためのアデノ随伴ウイルスベクターバリアント及びその方法
WO2018191440A1 (fr) Édition génomique in vivo de progéniteurs sanguins
Wang et al. Curative in vivo hematopoietic stem cell gene therapy of murine thalassemia using large regulatory elements
EP3177313B1 (fr) Thérapies à base de transcriptase inverse de la télomérase pour le traitement d&#39;états d&#39;associés à l&#39;infarctus du myocarde
US11268107B2 (en) SMAD7 gene delivery into muscle cells
JP7469328B2 (ja) 遺伝子操作された造血幹細胞によるベータサラセミア表現型の補正
JP2023507174A (ja) Dmd変異の修正のための方法及び組成物
Ramamurthy et al. Comparison of different gene addition strategies to modify placental derived-mesenchymal stromal cells to produce FVIII
JP2021522858A (ja) 心臓、骨格筋、及び筋幹細胞におけるインビボ相同組換え修復
US20210180087A1 (en) In situ gene editing
WO2023244737A1 (fr) Amplificateurs et vecteurs améliorés
WO2023220364A2 (fr) Méthodes et compositions améliorées pour la distribution de transgène et/ou la reconstitution de microglie
KR20240027748A (ko) Rbm20 돌연변이의 게놈 편집
Gutiérrez-Guerrero Gene Editing as an Alternative to Retroviral Vectors for Wiskott-Aldrich syndrome Gene therapy

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 3193943

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3485914

Country of ref document: EP

Kind code of ref document: P

Ref document number: 3848056

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20221006

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20231004

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
INTG Intention to grant announced

Effective date: 20240301

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20240513